Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
BMC Bioinformatics ; 21(Suppl 10): 354, 2020 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-32838732

RESUMO

BACKGROUND: Type A influenza viruses circulate and spread among wild birds and mostly consist of low pathogenic strains. However, fast genome variation timely results in the insurgence of high pathogenic strains, which when infecting poultry birds may cause a million deaths and strong commercial damage. More importantly, the host shift may concern these viruses and sustained human-to-human transmission may result in a dangerous pandemic outbreak. Therefore, fingerprints specific to either low or high pathogenic strains may represent a very important tool for global surveillance. RESULTS: We combined Normal Modes Analysis and surface electrostatic analysis of a mixed strain dataset of influenza A virus haemagglutinins from high and low pathogenic strains in order to infer specific fingerprints. Normal Modes Analysis sorted the strains in two different, homogeneous clusters; sorting was independent of clades and specific instead to high vs low pathogenicity. A deeper analysis of fluctuations and flexibility regions unveiled a special role for the 110-helix region. Specific sorting was confirmed by surface electrostatics analysis, which further allowed to focus on regions and mechanisms possibly crucial to the low-to-high transition. CONCLUSIONS: Evidence from previous work demonstrated that changes in surface electrostatics are associated with the evolution and spreading of avian influenza A virus clades, and seemingly involved also in the avian to mammalian host shift. This work shows that a combination of electrostatics and Normal Modes Analysis can also identify fingerprints specific to high and low pathogenicity. The possibility to predict which specific mutations may result in a shift to high pathogenicity may help in surveillance and vaccine development.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Vírus da Influenza A/metabolismo , Vírus da Influenza A/patogenicidade , Eletricidade Estática , Algoritmos , Animais , Animais Selvagens/virologia , Aves/virologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Influenza Aviária/virologia , Modelos Moleculares , Domínios Proteicos
2.
J Gen Virol ; 101(8): 816-824, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31855133

RESUMO

Neuraminidase inhibitors (NAIs) are the gold standard treatment for influenza A virus (IAV). Oseltamivir is mostly used, followed by zanamivir (ZA). NAIs are not readily degraded in conventional wastewater treatment plants and can be detected in aquatic environments. Waterfowl are natural IAV hosts and replicating IAVs could thus be exposed to NAIs in the environment and develop resistance. Avian IAVs form the genetic basis for new human IAVs, and a resistant IAV with pandemic potential poses a serious public health threat, as NAIs constitute a pandemic preparedness cornerstone. Resistance development in waterfowl IAVs exposed to NAIs in the water environment has previously been investigated in an in vivo mallard model and resistance development was demonstrated in several avian IAVs after the exposure of infected ducks to oseltamivir, and in an H1N1 IAV after exposure to ZA. The N1 and N2 types of IAVs have different characteristics and resistance mutations, and so the present study investigated the exposure of an N2-type IAV (H4N2) in infected mallards to 1, 10 and 100 µg l-1 of ZA in the water environment. Two neuraminidase substitutions emerged, H274N (ZA IC50 increased 5.5-fold) and E119G (ZA IC50 increased 110-fold) at 10 and 100 µg l-1 of ZA, respectively. Reversion towards wild-type was observed for both substitutions in experiments with removed drug pressure, indicating reduced fitness of both resistant viruses. These results corroborate previous findings that the development of resistance to ZA in the environment seems less likely to occur than the development of resistance to oseltamivir, adding information that is useful in planning for prudent drug use and pandemic preparedness.


Assuntos
Anseriformes/virologia , Farmacorresistência Viral/efeitos dos fármacos , Vírus da Influenza A/efeitos dos fármacos , Influenza Aviária/tratamento farmacológico , Oseltamivir/farmacologia , Zanamivir/farmacologia , Animais , Antivirais/farmacologia , Patos/virologia , Vírus da Influenza A/genética , Influenza Aviária/virologia , Mutação/efeitos dos fármacos
3.
BMC Vet Res ; 16(1): 432, 2020 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-33167987

RESUMO

BACKGROUND: South Korea conducts annual national surveillance programs to detect avian influenza (AI) in domestic poultry, live bird markets, and wild birds. In March 2017, an AIV was isolated from fecal samples in an outdoor aviary flight cage in a zoo in Korea. RESULTS: Nucleotide sequencing identified the isolate as low pathogenic avian influenza virus (LPAIV) H7N7, and DNA barcoding analysis identified the host species as red-crowned crane. This isolate was designated A/red-crowned crane/Korea/H1026/2017 (H7N7). Genetic analysis and gene constellation analysis revealed that A/red-crowned crane/Korea/H1026/2017 (H7N7) showed high similarity with four H7N7 LPAIVs isolated from wild bird habitats in Seoul and Gyeonggi in early 2017. CONCLUSIONS: Considering the genetic similarity and similar collection dates of the viruses, and the fact that zoo bird cages are vulnerable to AIV, it is likely that fecal contamination from wild birds might have introduced LPAIV H7N7 into the red-crowned crane at the zoo. Therefore, our results emphasize that enhanced biosecurity measures should be employed during the wild bird migration season, and that continued surveillance should be undertaken to prevent potential threats to avian species in zoos and to humans.


Assuntos
Vírus da Influenza A Subtipo H7N7/isolamento & purificação , Influenza Aviária/virologia , Animais , Animais de Zoológico/virologia , Aves , Fezes/virologia , Vírus da Influenza A Subtipo H7N7/genética , República da Coreia
4.
Avian Pathol ; 48(2): 98-110, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30484684

RESUMO

An outbreak of low pathogenic avian influenza (LPAI) subtype H6N1 (intravenous pathogenicity index = 0.11) infection occurred in four productive brown layer flocks on three farms in the Netherlands within a period of two months. The farms were located at a maximum distance of 4.6 km from each other. The infections were associated with egg production drops up to 74%, pale eggshells and persisting high mortality up to 3.2% per week. Three flocks were slaughtered prematurely as they were not profitable anymore. Newcastle disease, infectious bronchitis, egg drop syndrome and Mycoplasma gallisepticum infections could very likely be excluded as cause of or contributor to the condition in the field. Also, the anticoccidial drug nicarbazin, which can cause egg production drops and eggshell decolouration, was not detected in eggs from affected flocks. Furthermore, post mortem examinations revealed no lesions indicative of bacterial infection. Moreover, bacteriological analysis of hens was negative. The condition was reproduced in commercial brown layers after intratracheal inoculation with virus isolates from affected flocks. It is concluded that the LPAI H6N1 virus is very likely the only cause of the disease. An overview of main manuscripts published since 1976 describing non-H5 and non-H7 avian influenza (AI) virus infections in chickens and their biological significance is included in the present study, in which once more is shown that not only high pathogenic AI virus subtypes H5 and H7 can be detrimental to flocks of productive layers, but also non-H5 and non-H7 LPAI viruses (H6N1 virus). RESEARCH HIGHLIGHTS LPAI H6N1 can be detrimental to productive layers Detrimental effects are severe egg drop and persistent high mortality LPAI H6N1 virus outbreak seems to be self-limiting.


Assuntos
Galinhas/virologia , Surtos de Doenças/veterinária , Vírus da Influenza A/patogenicidade , Influenza Aviária/epidemiologia , Doenças das Aves Domésticas/epidemiologia , Animais , Casca de Ovo/patologia , Ovos , Feminino , Influenza Aviária/mortalidade , Influenza Aviária/patologia , Influenza Aviária/virologia , Países Baixos/epidemiologia , Doenças das Aves Domésticas/mortalidade , Doenças das Aves Domésticas/patologia , Doenças das Aves Domésticas/virologia
6.
Emerg Infect Dis ; 24(1): 149-152, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29260672

RESUMO

Infections with low pathogenicity and highly pathogenic avian influenza A(H7N9) viruses affected poultry in 4 states in the southeastern United States in 2017. We evaluated pathogenicity and transmission of representative viruses in mouse and ferret models and examined replication kinetics in human respiratory tract cells. These viruses can cause respiratory infections in mammalian models.


Assuntos
Subtipo H7N9 do Vírus da Influenza A/patogenicidade , Influenza Aviária/virologia , Infecções por Orthomyxoviridae/veterinária , Animais , Linhagem Celular , Galinhas/virologia , Surtos de Doenças/veterinária , Furões/virologia , Humanos , Influenza Aviária/epidemiologia , Influenza Humana/virologia , Camundongos , Infecções por Orthomyxoviridae/virologia , Sistema Respiratório/citologia , Tennessee/epidemiologia , Virulência
7.
8.
Avian Pathol ; 46(5): 488-496, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28417679

RESUMO

Field observations indicate that the impact of velogenic Newcastle disease virus (vNDV) is more severe in countries with concomitant circulation of low pathogenicity avian influenza virus, as is the case in the Middle East, in particular in Israel, where H9N2 and NDV are endemic. In our study, we evaluated how the exposure of chickens to an H9N2 challenge either favours or interferes with a subsequent vNDV infection and its transmission to sentinels. For this purpose, single vNDV and sequential H9/NDV challenges were performed with increasing doses of vNDV (101-106 EID50). The H9N2 challenge made birds more susceptible to the vNDV, lowering the minimum dose required to cause an infection, exacerbating the clinical outcome, while delaying the onset of the disease and time of death. Interestingly, the presence and degree of these seemingly contrasting effects were dose-dependent and not mutually exclusive.


Assuntos
Galinhas , Vírus da Influenza A Subtipo H9N2 , Influenza Aviária/virologia , Doença de Newcastle/virologia , Vírus da Doença de Newcastle , Doenças das Aves Domésticas/virologia , Animais , Anticorpos Antivirais , Coinfecção/veterinária , Organismos Livres de Patógenos Específicos
9.
Avian Dis ; 59(2): 344-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26473689

RESUMO

In April 2014 an outbreak of low pathogenic avian influenza H5N8 North American genetic lineage was diagnosed in a commercial quail operation in Stanislaus County, California. Sudden increase in mortality prompted the submission of 20 Japanese quail hens (Coturnix c. japonica) to the California Animal Health and Food Safety Laboratory, Turlock Branch. Oropharyngeal and cloacal swabs tested positive for influenza A virus H5N8 by real-time reverse transcription-polymerase chain reaction. The virus was subsequently isolated. In vivo assay and sequencing of the hemagglutinin protein cleavage site classified the virus as a North American genetic lineage of low pathogenicity for chickens. Following the diagnosis, a rapid and coordinated response took place to contain the outbreak. The affected premise was depopulated, cleaned, and disinfected. Three areas from the affected premises-a 3 kilometer (km) radius (High Risk Zone), a 3-10 km area (Buffer Zone), and a 10-20 km (Surveillance Zone)-were established for avian influenza testing of commercial and noncommercial poultry operations. Surveillance testing and rapid control measures were successful in the control and eradication of the outbreak and revealed no area of spread of the virus from the index flock. This report describes the history, diagnosis, surveillance, and control measures applied to manage this outbreak.


Assuntos
Coturnix , Vírus da Influenza A/classificação , Influenza Aviária/virologia , Doenças das Aves Domésticas/virologia , Animais , California/epidemiologia , Influenza Aviária/epidemiologia , Doenças das Aves Domésticas/epidemiologia
10.
Viruses ; 16(2)2024 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-38400060

RESUMO

Avian influenza virus (AIV) is a pathogen with zoonotic and pandemic potential. Migratory birds are natural reservoirs of all known subtypes of AIVs, except for H17N10 and H18N11, and they have been implicated in previous highly pathogenic avian influenza outbreaks worldwide. This study identified and characterized the first isolate of the H13N6 subtype from a Vega gull (Larus vegae mongolicus) in South Korea. The amino acid sequence of hemagglutinin gene showed a low pathogenic AIV subtype and various amino acid substitutions were found in the sequence compared to the reference sequence and known H13 isolates. High sequence homology with other H13N6 isolates was found in HA, NA, PB1, and PA genes, but not for PB2, NP, M, and NS genes. Interestingly, various point amino acid mutations were found on all gene segments, and some are linked to an increased binding to human-type receptors, resistance to antivirals, and virulence. Evolutionary and phylogenetic analyses showed that all gene segments are gull-adapted, with a phylogeographic origin of mostly Eurasian, except for PB2, PA, and M. Findings from this study support the evidence that reassortment of AIVs continuously occurs in nature, and migratory birds are vital in the intercontinental spread of avian influenza viruses.


Assuntos
Charadriiformes , Vírus da Influenza A , Influenza Aviária , Animais , Humanos , Filogenia , Aves
11.
Am J Vet Res ; 85(5)2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38593825

RESUMO

Highly pathogenic avian influenza (HPAI) has persisted as a One Health threat whose current circulation and impact are addressed in the companion Currents in One Health by Puryear and Runstadler, JAVMA, May 2024. Highly pathogenic avian influenza emerged as a by-product of agricultural practices and adapted to endemic circulation in wild bird species. Over more than 20 years, continued evolution in a complex ecology involving multiple hosts has produced a lineage that expanded globally over the last 2 years. Understanding the continued evolution and movement of HPAI relies on understanding how the virus is infecting different hosts in different contexts. This includes understanding the environmental factors and the natural ecology of viral transmission that impact host exposure and ultimately evolutionary trajectories. Particularly with the rapid host expansion, increased spillover to mammalian hosts, and novel clinical phenotypes in infected hosts, despite progress in understanding the impact of specific mutations to HPAI viruses that are associated with spillover potential, the threat to public health is poorly understood. Active research is focusing on new approaches to understanding the relationship of viral genotype to phenotype and the implementation of research and surveillance pipelines to make sense of the enormous potential for diverse HPAI viruses to emerge from wild reservoirs amid global circulation.


Assuntos
Animais Selvagens , Aves , Influenza Aviária , Mamíferos , Animais , Influenza Aviária/virologia , Influenza Aviária/transmissão , Influenza Aviária/epidemiologia , Animais Selvagens/virologia , Aves/virologia , Mamíferos/virologia , Infecções por Orthomyxoviridae/veterinária , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/transmissão , Infecções por Orthomyxoviridae/epidemiologia , Vírus da Influenza A/patogenicidade , Vírus da Influenza A/genética , Doenças Transmissíveis Emergentes/virologia , Doenças Transmissíveis Emergentes/veterinária , Doenças Transmissíveis Emergentes/transmissão
12.
Poult Sci ; 103(10): 104089, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-39142030

RESUMO

Avian chlamydiosis is a serious avian infection that carries a significant zoonotic danger to the poultry industry. The respiratory co-infections caused by the low pathogenic avian influenza virus H9N2 (LPAIV H9N2) also cause significant financial losses in the poultry industry. The purpose of this study was to examine the pathogenicity of Chlamydophila psittaci, and LPAIV H9N2 individually and in combination in broiler chickens, as well as to determine whether or not aqueous neem (Azadirachta indica) leaf extract is effective against infections caused by these pathogens. Therefore, 120 broiler cobb chicks were equally divided into 4 groups (30 birds each) with triplicates with 10 birds. Broilers in group 1 (G1) were infected with only C. psittaci, broilers in group 2 (G2) were infected with only LPAIV H9N2, broilers in group 3 (G3) were infected with C. psittaci and LPAIV H9N2, and broilers in group 4 (G4) remained not challenged and non-treated with any therapeutic or preventive treatment (negative control). At 21 d postinfection (dpi), birds in G1, G2, and G3 were divided into 3 subgroups of 10 birds each: subgroup (A) remained infected and untreated (positive control), subgroup (B) infected and received oxytetracycline for 5 consecutive d, and subgroup (C) infected and received 8% aqueous neem leaf extract for 5 consecutive d. The multiplication of C. psittaci in birds in G1, in various tissues was evaluated using Giemsa staining and the data showed that multiplication was much higher in the lung, spleen, and liver from 6 h to 21 dpi, but low in the heart from 8 to 21 dpi. During simultaneous co-infection in G3, the birds developed significant clinical symptoms and postmortem lesions (PM). Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect viral shedding from oropharyngeal and cloacal swabs between 2 dpi and 8 dpi, with cycle threshold (CT) values ranging from 22 to 24. In contrast, bacterial shedding began 6 h after infection and continued until 21 dpi, with CT values ranging from 23 to 26. Administration of an aqueous neem leaf extract at an 8% concentration (Group C) resulted in a numerical rise in average body weight across all treatment groups in the third and fourth week, as well as a reduction in LPAIV H9N2 and C. psittaci replication in the respiratory and gut of treated birds compared to those treated with oxytetracycline (Group B). Overall, respiratory co-infections pose a considerable risk to the poultry business, which is a big threat. To control C. psittaci and LPAIV H9N2 in broiler chickens, oral supplementation of 8% aqueous neem leaf extract is recommended. This treatment improves the birds' performance, as evidenced by an increase in their average body weight. In addition, the application of 8% aqueous neem leaf extract lowers C. psittaci replication within tissues and diminishes LPAIV H9N2 shedding.

13.
J Food Prot ; 87(8): 100325, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38964610

RESUMO

With the emergence of clade 2.3.4.4b H5N1 highly pathogenic avian influenza virus (AIV) infection of dairy cattle and its subsequent detection in raw milk, coupled with recent AIV infections affecting dairy farm workers, experiments were conducted to affirm the safety of cooked ground beef related to AIV because such meat is often derived from cull dairy cows. Specifically, retail ground beef (percent lean:fat = ca. 80:20) was inoculated with a low pathogenic AIV (LPAIV) isolate to an initial level of 5.6 log10 50% egg infectious doses (EID50)  per 300 g patty. The inoculated meat was pressed into patties (ca. 2.54 cm thick, ca. 300 g each) and then held at 4 °C for up to 60 min. In each of the two trials, two patties for each of the following three treatments were cooked on a commercial open-flame gas grill to internal instantaneous temperatures of 48.9 °C (120°F), 62.8 °C (145°F), or 71.1 °C (160°F), but without any dwell time. Cooking inoculated ground beef patties to 48.9 °C (ave. cooking time of ca. 15 min) resulted in a mean reduction of ≥2.5 ± 0.9 log10 EID50 per 300 g of ground beef as assessed via quantification of virus in embryonating chicken eggs (ECEs). Likewise, cooking patties on a gas grill to 62.8 °C (ave. cooking time of ca. 21 min) or to the USDA FSIS recommended minimum internal temperature for ground beef of 71.1 °C (ave. cooking time of ca. 24 min) resulted in a reduction to nondetectable levels from initial levels of ≥5.6 log10 EID50 per 300 g. These data establish that levels of infectious AIV are substantially reduced within inoculated ground beef patties (20% fat) using recommended cooking procedures.


Assuntos
Culinária , Animais , Bovinos , Humanos , Influenza Aviária , Carne Vermelha , Virus da Influenza A Subtipo H5N1 , Carne , Aves
14.
J Lipid Res ; 54(10): 2733-44, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23883582

RESUMO

Although HDL is inversely correlated with coronary heart disease, elevated HDL-cholesterol is not always protective. Additionally, HDL has biological functions that transcend any antiatherogenic role: shotgun proteomics show that HDL particles contain 84 proteins (latest count), many correlating with antioxidant and anti-inflammatory properties of HDL. ApoA-I has been suggested to serve as a platform for the assembly of these protein components on HDL with specific functions - the HDL proteome. However, the stoichiometry of apoA-I in HDL subspecies is poorly understood. Here we use a combination of immunoaffinity chromatography data and volumetric analysis to evaluate the size and stoichiometry of LpA-I and LpA-I,A-II particles. We conclude that there are three major LpA-I subspecies: two major particles, HDL[4] in the HDL3 size range (d = 85.0 ± 1.2 Å) and HDL[7] in the HDL2 size range (d = 108.5 ± 3.8 Å) with apoA-I stoichiometries of 3 and 4, respectively, and a small minor particle, HDL[1] (d = 73.8 ± 2.1Å) with an apoA-I stoichiometry of 2. Additionally, we conclude that the molar ratio of apolipoprotein to surface lipid is significantly higher in circulating HDL subspecies than in reconstituted spherical HDL particles, presumably reflecting a lack of phospholipid transfer protein in reconstitution protocols.


Assuntos
Apolipoproteína A-II/sangue , Apolipoproteína A-I/sangue , Lipoproteínas HDL/sangue , Cromatografia de Afinidade , Feminino , Humanos , Lipoproteína(a)/sangue , Lipoproteínas HDL/química , Lipoproteínas HDL/isolamento & purificação , Masculino , Eletroforese em Gel de Poliacrilamida Nativa , Tamanho da Partícula , Propriedades de Superfície , Ultracentrifugação
15.
Viruses ; 15(2)2023 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-36851676

RESUMO

Low-pathogenic avian influenza (LPAI) H9N2 virus is endemic in Bangladesh, causing huge economic losses in the poultry industry. Although a considerable number of Bangladeshi LPAI H9N2 viruses have been molecularly characterized, there is inadequate information on the pathogenicity of H9N2 viruses in commercial poultry. In this study, circulating LPAI H9N2 viruses from recent field outbreaks were characterized, and their pathogenicity in commercial Sonali (crossbred) and broiler chickens was assessed. Phylogenetic analysis of currently circulating field viruses based on the hemagglutinin (HA) and neuraminidase (NA) gene sequences revealed continuous circulation of G1 lineages containing the tri-basic hemagglutinin cleavage site (HACS) motif (PAKSKR*GLF) at the HA protein. Both the LPAI susceptible Sonali and broiler chickens were infected with selected H9N2 isolates A/chicken/Bangladesh/2458-LT2/2020 or A/chicken/Bangladesh/2465-LT56/2021 using intranasal (100 µL) and intraocular (100 µL) routes with a dose of 106 EID50/mL. Infected groups (LT_2-So1 and LT_56-So2; LT_2-Br1 and LT_56-Br2) revealed no mortality or clinical signs. However, at gross and histopathological investigation, the trachea, lungs, and intestine of the LT_2-So1 and LT_56-So2 groups displayed mild to moderate hemorrhages, congestion, and inflammation at different dpi. The LT 2-Br1 and LT 56-Br2 broiler groups showed nearly identical changes in the trachea, lungs, and intestine at various dpi, indicating no influence on pathogenicity in the two commercial bird species under study. Overall, the prominent lesions were observed up to 7 dpi and started to disappear at 10 dpi. The H9N2 viruses predominantly replicated in the respiratory tract, and higher titers of virus were shed through the oropharyngeal route than the cloacal route. Finally, this study demonstrated the continuous evolution of tri-basic HACS containing H9N2 viruses in Bangladesh with a low-pathogenic phenotype causing mild to moderate tracheitis, pneumonia, and enteritis in Sonali and commercial broiler chickens.


Assuntos
Vírus da Influenza A Subtipo H9N2 , Influenza Aviária , Animais , Galinhas , Vírus da Influenza A Subtipo H9N2/genética , Hemaglutininas , Filogenia , Virulência
16.
EFSA J ; 21(12): e8480, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38099051

RESUMO

All European Union (EU) Member States (MSs) are required to implement surveillance for avian influenza (AI) in poultry and wild birds and (i) to notify the outbreaks, when relevant and (ii) to report the results to the responsible authority. In addition, Iceland, Norway, Switzerland and the United Kingdom (Northern Ireland) also implement ongoing surveillance programmes to monitor occurrences of avian influenza viruses (AIVs) in poultry and wild birds. EFSA received a mandate from the European Commission to collate, validate, analyse and summarise the data resulting from these AI surveillance programmes in an annual report. The present report summarises the results of the surveillance activities carried out in MSs, Iceland, Norway, Switzerland and the United Kingdom (Northern Ireland) in 2022. Overall, the 31 reporting countries (RCs) sampled 22,171 poultry establishments (PEs) during the 2022 surveillance activity: 18,490 PEs were sampled for serological testing and 3775 were sampled for virological testing. Some PEs were therefore sampled for both type of analytical methods. Out of the 18,490 PEs sampled for serological testing, 15 (0.08%) were seropositive for influenza A(H5) viruses. Out of the 3775 PEs sampled for virological testing, 74 PEs (1.96%) were positive to the virological assay for influenza A(H5) viruses. Seropositive PEs were found in four RCs (Belgium, Poland, Spain and Sweden) and as in previous years, the highest percentages of seropositive PEs were found in PEs raising breeding geese and waterfowl game birds. Out of these 15 seropositive PEs, 3 also tested positive by polymerase chain reaction (PCR) for influenza A (H5) viruses - 2 for highly pathogenic avian influenza virus (HPAIV) and 1 low pathogenic avian influenza (LPAI) (H5N3). In relation to the virological surveys, 10 RCs (32%) out of the 31 reported the detection of A (H5) viruses in 74 PEs, covering 12 different poultry categories. More specifically, 54 reported HPAIV A(H5N1), 17 HPAIV (H5N8), 2 AIV (H5N1) with unknown virus pathogenicity and 1 low pathogenic avian influenza (LPAI) (H5N3). Additionally, six PEs tested positive for undefined AIVs in three RCs. A total of 32,143 wild birds were sampled, with 4163 (12.95%) wild birds testing positive for HPAIVs by PCR, from 25 RCs. In contrast to previous years, out of the 4163 wild birds testing positive for HPAIv, subtype A(H5N1) virus was the main influenza A virus subtype identified among the wild bird testing positive for HPAIVs (3942; 95%). In addition, RCs also reported 984 wild birds testing positive for low pathogenic avian influenza (LPAI). Out of those, for 660 (67%) it was ascertained that the subtype was non-A(H5/H7); 260 (26%) wild birds tested positive for LPAIv of A(H5 or H7) subtypes and the remaining 64 (7%) LPAI viruses were belonging to other H-subtypes.

17.
EFSA J ; 21(1): e07786, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36698491

RESUMO

Between October 2021 and September 2022 Europe has suffered the most devastating highly pathogenic avian influenza (HPAI) epidemic with a total of 2,520 outbreaks in poultry, 227 outbreaks in captive birds, and 3,867 HPAI virus detections in wild birds. The unprecedent geographical extent (37 European countries affected) resulted in 50 million birds culled in affected establishments. In the current reporting period, between 10 September and 2 December 2022, 1,163 HPAI virus detections were reported in 27 European countries in poultry (398), captive (151) and wild birds (613). A decrease in HPAI virus detections in colony-breeding seabirds species and an increase in the number of detections in waterfowl has been observed. The continuous circulation of the virus in the wild reservoir has led to the frequent introduction of the virus into poultry populations. It is suspected that waterfowl might be more involved than seabirds in the incursion of HPAI virus into poultry establishments. In the coming months, the increasing infection pressure on poultry establishments might increase the risk of incursions in poultry, with potential further spread, primarily in areas with high poultry densities. The viruses detected since September 2022 (clade 2.3.4.4b) belong to eleven genotypes, three of which have circulated in Europe during the summer months, while eight represent new genotypes. HPAI viruses were also detected in wild and farmed mammal species in Europe and North America, showing genetic markers of adaptation to replication in mammals. Since the last report, two A(H5N1) detections in humans in Spain, one A(H5N1), one A(H5N6) and one A(H9N2) human infection in China as well as one A(H5) infection without NA-type result in Vietnam were reported, respectively. The risk of infection is assessed as low for the general population in the EU/EEA, and low to medium for occupationally exposed people.

18.
Viruses ; 15(2)2023 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-36851517

RESUMO

Influenza A viruses are rarely symptomatic in wild birds, while representing a higher threat to poultry and mammals, where they can cause a variety of symptoms, including death. H5 and H7 subtypes of influenza viruses are of particular interest because of their pathogenic potential and reported capacity to spread from poultry to mammals, including humans. The identification of molecular fingerprints for pathogenicity can help surveillance and early warning systems, which are crucial to prevention and protection from such potentially pandemic agents. In the past decade, comparative analysis of the surface features of hemagglutinin, the main protein antigen in influenza viruses, identified electrostatic fingerprints in the evolution and spreading of H5 and H9 subtypes. Electrostatic variation among viruses from avian or mammalian hosts was also associated with host jump. Recent findings of fingerprints associated with low and highly pathogenic H5N1 viruses, obtained by means of comparative electrostatics and normal modes analysis, prompted us to check whether such fingerprints can also be found in the H7 subtype. Indeed, evidence presented in this work showed that also in H7N7, hemagglutinin proteins from low and highly pathogenic strains present differences in surface electrostatics, while no meaningful variation was found in normal modes.


Assuntos
Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A Subtipo H7N7 , Influenza Aviária , Animais , Humanos , Hemaglutininas , Eletricidade Estática , Mamíferos
19.
Microbiol Spectr ; 10(1): e0082221, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35019707

RESUMO

The H9N2 subtype avian influenza virus (AIV) has become endemic in poultry globally; however due to its low pathogenicity, it is not under primary surveillance and control in many countries. Recent reports of human infection caused by H9N2 AIV has increased public concern. This study investigated the genetic and antigenic characteristics of H9N2 AIV isolated from local markets in nine provinces in Southern China from 2013 to 2018. We detected an increasing annual isolation rate of H9N2 AIV. Phylogenetic analyses of hemagglutinin (HA) genes suggests that isolated strains were rooted in BJ94 lineage but have evolved into new subgroups (II and III), which derived from subgroup I. The estimated substitution rate of the subgroup III strains was 6.23 × 10-3 substitutions/site/year, which was 1.5-fold faster than that of the average H9N2 HA rate (3.95 × 10-3 substitutions/site/year). Based on the antigenic distances, subgroup II and III strains resulted in two clear antigenic clusters 2 and 3, separated from the vaccine strain F98, cluster 1. New antigenic properties of subgroup III viruses were associated with 11 amino acid changes in the HA protein, suggesting antigenic drift in H9N2 viruses. Our phylogenetic and antigenic analyses of the H9N2 strains circulating in local markets in Southern China provide new insights on the antigenic diversification of H9N2 viruses. IMPORTANCE The H9N2 low pathogenicity avian influenza (LPAI) virus has become endemic in poultry globally. In several Asian countries, vaccination against H9N2 avian influenza virus (AIV) was approved to reduce economic losses in the poultry industry. However, surveillance programs initiated after the introduction of vaccination identified the persistence of H9N2 AIV in poultry (especially in chicken in South Korea and China). Recent reports of human infection caused by H9N2 AIV has increased public concern. Surveillance of H9N2 circulating in poultry in the fields or markets was essential to update the vaccination strategies. This study investigated the genetic and antigenic characteristics of H9N2 AIVs isolated from local markets in nine provinces in Southern China from 2013 to 2018. The discovery of mutations in the hemagglutinin (HA) gene that result in antigenic changes provides a baseline reference for evolutionary studies of H9N2 viruses and vaccination strategies in poultry.


Assuntos
Evolução Molecular , Vírus da Influenza A Subtipo H9N2/genética , Vírus da Influenza A Subtipo H9N2/imunologia , Influenza Aviária/virologia , Doenças das Aves Domésticas/virologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Deriva e Deslocamento Antigênicos , Variação Antigênica , Galinhas , China/epidemiologia , Hemaglutininas Virais/química , Hemaglutininas Virais/genética , Hemaglutininas Virais/imunologia , Vírus da Influenza A Subtipo H9N2/classificação , Vírus da Influenza A Subtipo H9N2/isolamento & purificação , Influenza Aviária/epidemiologia , Filogenia , Doenças das Aves Domésticas/epidemiologia
20.
EFSA J ; 20(9): e07554, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36177389

RESUMO

European Union (EU) Member States (MSs) are required to carry out surveillance for avian influenza (AI) in poultry and wild birds and notify the results to the responsible authority. In addition, Iceland, Norway, Switzerland and the United Kingdom (Northern Ireland) also implement ongoing surveillance programmes to monitor incursions of avian influenza viruses (AIVs) in poultry and wild birds. EFSA received a mandate from the European Commission to collate, validate, analyse and summarise the data resulting from these AI surveillance programmes in an annual report. The present report summarises the results of the surveillance activities carried out in MSs and the aforementioned countries in 2021. Overall, 24,290 poultry establishments (PEs) were sampled, of which 27 were seropositive for influenza A(H5) and 4 for A(H7) viruses. Seropositive PEs were found in 10 MSs and, as per previous years, the highest percentages of seropositive PEs were found in establishments raising waterfowl game birds and breeding geese. Out of these 31 seropositive PEs, 3 tested positive by polymerase chain reaction (PCR) for influenza A(H5) viruses: 1 for highly pathogenic avian influenza virus (HPAIV), 1 for low pathogenic avian influenza virus (LPAIV) and 1 with unknown virus pathogenicity. In addition, 16 countries reported PCR test results from 1,858 PEs which did not correspond to the follow-up testing of a positive serology event (e.g. in some PEs, PCR tests were used for screening). Sixty-five of these PEs in 10 MSs were found positive for AIVs. Apart from poultry, 31,382 wild birds were sampled, with 2,314 wild birds testing positive for HPAIVs by PCR. Twenty-two countries reported HPAIV-positive wild birds and most positive samples were identified as highly pathogenic avian influenza (HPAI) A(H5N8) virus. In addition, 328 wild birds tested positive for LPAIVs of the A(H5/H7) subtypes and 362 wild birds tested positive for non-A(H5/H7) subtype AIVs.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa